Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jaimee R. Compton is active.

Publication


Featured researches published by Jaimee R. Compton.


Biochemistry | 2012

Probing the Donor and Acceptor Substrate Specificity of the γ-Glutamyl Transpeptidase

Xin Hu; Patricia M. Legler; Ilja V. Khavrutskii; Angelo Scorpio; Jaimee R. Compton; Kelly L. Robertson; Arthur M. Friedlander; Anders Wallqvist

γ-Glutamyl transpeptidase (GGT) is a two-substrate enzyme that plays a central role in glutathione metabolism and is a potential target for drug design. GGT catalyzes the cleavage of γ-glutamyl donor substrates and the transfer of the γ-glutamyl moiety to an amine of an acceptor substrate or water. Although structures of bacterial GGT have revealed details of the protein-ligand interactions at the donor site, the acceptor substrate site is relatively undefined. The recent identification of a species-specific acceptor site inhibitor, OU749, suggests that these inhibitors may be less toxic than glutamine analogues. Here we investigated the donor and acceptor substrate preferences of Bacillus anthracis GGT (CapD) and applied computational approaches in combination with kinetics to probe the structural basis of the enzymes substrate and inhibitor binding specificities and compare them with human GGT. Site-directed mutagenesis studies showed that the R432A and R520S variants exhibited 6- and 95-fold decreases in hydrolase activity, respectively, and that their activity was not stimulated by the addition of the l-Cys acceptor substrate, suggesting an additional role in acceptor binding and/or catalysis of transpeptidation. Rat GGT (and presumably HuGGT) has strict stereospecificity for L-amino acid acceptor substrates, while CapD can utilize both L- and D-acceptor substrates comparably. Modeling and kinetic analysis suggest that R520 and R432 allow two alternate acceptor substrate binding modes for L- and D-acceptors. R432 is conserved in Francisella tularensis, Yersinia pestis, Burkholderia mallei, Helicobacter pylori and Escherichia coli, but not in human GGT. Docking and MD simulations point toward key residues that contribute to inhibitor and acceptor substrate binding, providing a guide to designing novel and specific GGT inhibitors.


Proteins | 2011

Introduction of a disulfide bond leads to stabilization and crystallization of a ricin immunogen.

Jaimee R. Compton; Patricia M. Legler; Benjamin V. Clingan; Mark A. Olson; Charles B. Millard

RTA1‐33/44‐198 is a catalytically inactive, single‐domain derivative of the ricin toxin A‐chain (RTA) engineered to serve as a stable protein scaffold for presentation of native immunogenic epitopes (Olson et al., Protein Eng Des Sel 2004;17:391–397). To improve the stability and solubility of RTA1‐33/44‐198 further, we have undertaken the design challenge of introducing a disulfide (SS) bond. Nine pairs of residues were selected for placement of the SS‐bond based on molecular dynamics simulation studies of the modeled single‐domain chain. Disulfide formation at either of two positions (R48C/T77C or V49C/E99C) involving a specific surface loop (44–55) increased the protein melting temperature by ∼5°C compared with RTA1‐33/44‐198 and by ∼13°C compared with RTA. Prolonged stability studies of the R48C/T77C variant (>60 days at 37°C, pH 7.4) confirmed a >40% reduction in self‐aggregation compared with RTA1‐33/44‐198 lacking the SS‐bond. The R48C/T77C variant retained affinity for anti‐RTA antibodies capable of neutralizing ricin toxin, including a monoclonal that recognizes a human B‐cell epitope. Introduction of either R48C/T77C or V49C/E99C promoted crystallization of RTA1‐33/44‐198, and the X‐ray structures of the variants were solved to 2.3Å or 2.1 Å resolution, respectively. The structures confirm formation of an intramolecular SS‐bond, and reveal a single‐domain fold that is significantly reduced in volume compared with RTA. Loop 44 to 55 is partly disordered as predicted by simulations, and is positioned to form self‐self interactions between symmetry‐related molecules. We discuss the importance of RTA loop 34 to 55 as a nucleus for unfolding and aggregation, and draw conclusions for ongoing structure‐based minimalist design of RTA‐based immunogens. Proteins 2011. Published 2010 Wiley‐Liss, Inc.


Proteins | 2014

Structural and mutational analysis of a monomeric and dimeric form of a single domain antibody with implications for protein misfolding.

Jade George; Jaimee R. Compton; Dagmar H. Leary; Mark A. Olson; Patricia M. Legler

Camelid single domain antibodies (sdAb) are known for their thermal stability and reversible refolding. We have characterized an unusually stable sdAb recognizing Staphylococcal enterotoxin B with one of the highest reported melting temperatures (Tm = 85°C). Unexpectedly, ∼10−20% of the protein formed a dimer in solution. Three other cases where <20% of the sdAb dimerized have been reported; however, this is the first report of both the monomeric and dimeric X‐ray crystal structures. Concentration of the monomer did not lead to the formation of new dimer suggesting a stable conformationally distinct species in a fraction of the cytoplasmically expressed protein. Comparison of periplasmic and cytoplasmic expression showed that the dimer was associated with cytoplasmic expression. The disulfide bond was partially reduced in the WT protein purified from the cytoplasm and the protein irreversibly unfolded. Periplasmic expression produced monomeric protein with a fully formed disulfide bond and mostly reversible refolding. Crystallization of a disulfide‐bond free variant, C22A/C99V, purified from the periplasm yielded a structure of a monomeric form, while crystallization of C22A/C99V from the cytoplasm produced an asymmetric dimer. In the dimer, a significant conformational asymmetry was found in the loop residues of the edge β‐strands (S50‐Y60) containing the highly variable complementarity determining region, CDR2. Two dimeric assemblies were predicted from the crystal packing. Mutation of a residue at one of the interfaces, Y98A, disrupted the dimer in solution. The pleomorphic homodimer may yield insight into the stability of misfolded states and the importance of the conserved disulfide bond in preventing their formation. Proteins 2014; 82:3101–3116.


Journal of Medicinal Chemistry | 2013

3-substituted indole inhibitors against Francisella tularensis FabI identified by structure-based virtual screening.

Xin Hu; Jaimee R. Compton; Mohamed Diwan M. AbdulHameed; Kelly L. Robertson; Dagmar H. Leary; Ajit Jadhav; Jeremy R. Hershfield; Anders Wallqvist; Arthur M. Friedlander; Patricia M. Legler

In this study, we describe novel inhibitors against Francisella tularensis SchuS4 FabI identified from structure-based in silico screening with integrated molecular dynamics simulations to account for induced fit of a flexible loop crucial for inhibitor binding. Two 3-substituted indoles, 54 and 57, preferentially bound the NAD(+) form of the enzyme and inhibited growth of F. tularensis SchuS4 at concentrations near that of their measured Ki. While 57 was species-specific, 54 showed a broader spectrum of growth inhibition against F. tularensis , Bacillus anthracis , and Staphylococcus aureus . Binding interaction analysis in conjunction with site-directed mutagenesis revealed key residues and elements that contribute to inhibitor binding and species specificity. Mutation of Arg-96, a poorly conserved residue opposite the loop, was unexpectedly found to enhance inhibitor binding in the R96G and R96M variants. This residue may affect the stability and closure of the flexible loop to enhance inhibitor (or substrate) binding.


Journal of Cellular Biochemistry | 2005

Histone acetylase inhibitor trichostatin A induces acetylcholinesterase expression and protects against organophosphate exposure.

Bryan F. Curtin; Lauren M. Tetz; Jaimee R. Compton; Bhupendra P. Doctor; Richard K. Gordon; Madhusoodana P. Nambiar

The biological effects of organophosphorous (OP) chemical warfare nerve agents (CWNAs) are exerted by inhibition of acetylcholinesterase (AChE), which prevents the hydrolysis of the neurotransmitter acetylcholine, leading to hypercholinergy, seizures/status epilepticus, respiratory/cardiovascular failure, and potentially death. Current investigations show that bioscavenger therapy using purified fetal bovine AChE in rodents and non‐human primates and the more recently tested human butyrylcholinesterase, is a promising treatment for protection against multiple LD50 CWNA exposures. Potential impediments, due to the complex structure of the enzyme, purification effort, resources, and cost have necessitated alternative approaches. Therefore, we investigated the effects of transcriptional inducers to enhance the expression of AChE to achieve sufficient protection against OP poisoning. Trichostatin A (TSA), an inhibitor of histone deacetylase that de‐condenses the chromatin, thereby increasing the binding of transcription factors and mRNA synthesis, was evaluated for induction of AChE expression in various neuronal cell lines. Dose‐response curves showed that a concentration of 333 nM TSA was optimal in inducing AChE expression. In Neuro‐2A cells, TSA at 333 nM increased the extracellular AChE activity approximately 3–4 fold and intracellular enzyme activity 10‐fold. Correlating with the AChE induction, TSA pre‐treatment significantly protected the cells against exposure to the organophosphate diisopropylfluorophosphate, a surrogate for the chemical warfare agents soman and sarin. These studies indicate that transcriptional inducers such as TSA up‐regulate AChE, which then can bioscavenge any organophosphates present, thereby protecting the cells from OP‐induced cytotoxicity. In conclusion, transcriptional inducers are prospective new methods to protect against CWNA exposure.


Biochemistry | 2016

Kinetic, Mutational, and Structural Studies of the Venezuelan Equine Encephalitis Virus Nonstructural Protein 2 Cysteine Protease

Xin Hu; Jaimee R. Compton; Dagmar H. Leary; Mark A. Olson; Michael S. Lee; Jonah Cheung; Wenjuan Ye; Mark Ferrer; Noel Southall; Ajit Jadhav; Elaine M. Morazzani; Pamela J. Glass; Juan J. Marugan; Patricia M. Legler

The Venezuelan equine encephalitis virus (VEEV) nonstructural protein 2 (nsP2) cysteine protease (EC 3.4.22.-) is essential for viral replication and is involved in the cytopathic effects (CPE) of the virus. The VEEV nsP2 protease is a member of MEROPS Clan CN and characteristically contains a papain-like protease linked to an S-adenosyl-l-methionine-dependent RNA methyltransferase (SAM MTase) domain. The protease contains an alternative active site motif, (475)NVCWAK(480), which differs from papains (CGS(25)CWAFS), and the enzyme lacks a transition state-stabilizing residue homologous to Gln-19 in papain. To understand the roles of conserved residues in catalysis, we determined the structure of the free enzyme and the first structure of an inhibitor-bound alphaviral protease. The peptide-like E64d inhibitor was found to bind beneath a β-hairpin at the interface of the SAM MTase and protease domains. His-546 adopted a conformation that differed from that found in the free enzyme; one or both of the conformers may assist in leaving group departure of either the amine or Cys thiolate during the catalytic cycle. Interestingly, E64c (200 μM), the carboxylic acid form of the E64d ester, did not inhibit the nsP2 protease. To identify key residues involved in substrate binding, a number of mutants were analyzed. Mutation of the motif residue, N475A, led to a 24-fold reduction in kcat/Km, and the conformation of this residue did not change after inhibition. N475 forms a hydrogen bond with R662 in the SAM MTase domain, and the R662A and R662K mutations both led to 16-fold decreases in kcat/Km. N475 forms the base of the P1 binding site and likely orients the substrate for nucleophilic attack or plays a role in product release. An Asn homologous to N475 is similarly found in coronaviral papain-like proteases (PLpro) of the Severe Acute Respiratory Syndrome (SARS) virus and Middle East Respiratory Syndrome (MERS) virus. Mutation of another motif residue, K480A, led to a 9-fold decrease in kcat and kcat/Km. K480 likely enhances the nucleophilicity of the Cys. Consistent with our substrate-bound models, the SAM MTase domain K706A mutation increased Km 4.5-fold to 500 μM. Within the β-hairpin, the N545A mutation slightly but not significantly increased kcat and Km. The structures and identified active site residues may facilitate the discovery of protease inhibitors with antiviral activity.


mAbs | 2017

Stability of isolated antibody-antigen complexes as a predictive tool for selecting toxin neutralizing antibodies.

Patricia M. Legler; Jaimee R. Compton; Martha L. Hale; George P. Anderson; Mark A. Olson; Charles B. Millard; Ellen R. Goldman

ABSTRACT Ricin is an A-B ribosome inactivating protein (RIP) toxin composed of an A-chain subunit (RTA) that contains a catalytic N-glycosidase and a B-chain (RTB) lectin domain that binds cell surface glycans. Ricin exploits retrograde transport to enter into the Golgi and the endoplasmic reticulum, and then dislocates into the cytoplasm where it can reach its substrate, the rRNA. A subset of isolated antibodies (Abs) raised against the RTA subunit protect against ricin intoxication, and RTA-based vaccine immunogens have been shown to provide long-lasting protective immunity against the holotoxin. Anti-RTA Abs are unlikely to cross a membrane and reach the cytoplasm to inhibit the enzymatic activity of the A-chain. Moreover, there is not a strict correlation between the apparent binding affinity (Ka) of anti-RTA Abs and their ability to successfully neutralize ricin toxicity. Some anti-RTA antibodies are toxin-neutralizing, whereas others are not. We hypothesize that neutralizing anti-RTA Abs may interfere selectively with conformational change(s) or partial unfolding required for toxin internalization. To test this hypothesis, we measured the melting temperatures (Tm) of neutralizing single-domain Ab (sdAb)-antigen (Ag) complexes relative to the Tm of the free antigen (Tm-shift = Tmcomplex – TmAg), and observed increases in the Tmcomplex of 9–20 degrees. In contrast, non-neutralizing sdAb-Ag complexes shifted the TmComplex by only 6–7 degrees. A strong linear correlation (r2 = 0.992) was observed between the magnitude of the Tm-shift and the viability of living cells treated with the sdAb and ricin holotoxin. The Tm-shift of the sdAb-Ag complex provided a quantitative biophysical parameter that could be used to predict and rank-order the toxin-neutralizing activities of Abs. We determined the first structure of an sdAb-RTA1-33/44-198 complex, and examined other sdAb-RTA complexes. We found that neutralizing sdAb bound to regions involved in the early stages of unfolding. These Abs likely interfere with steps preceding or following endocytosis that require conformational changes. This method may have utility for the characterization or rapid screening of other Ab that act to prevent conformational changes or unfolding as part of their mechanism of action.


FEMS Microbiology Ecology | 2015

A novel Vibrio beta-glucosidase (LamN) that hydrolyzes the algal storage polysaccharide laminarin

Zheng Wang; Kelly L. Robertson; Charles Liu; Jinny L. Liu; Brandy J. Johnson; Dagmar H. Leary; Jaimee R. Compton; Varaporn Vuddhakul; Patricia M. Legler; Gary J. Vora

The metabolic versatility, tractability and rapid growth potential of the Vibrio spp. have made them increasingly attractive systems for investigating carbon cycling in the marine environment. In this study, an in silico subtractive proteomic strategy was used to identify a novel 101 kDa GH3 family β-glucosidase (LamN) that was found in bioluminescent Vibrio campbellii strains capable of utilizing the algal storage glucan laminarin. A heterologous overexpression system verified the sequence-predicted function of LamN as it enabled the growth of Escherichia coli on laminarin as a sole carbon source. Quantitative reverse transcription PCR analyses revealed that V. campbellii grown on laminarin demonstrated a 4- to 314-fold induction of lamN gene expression when compared to the same strains grown on glucose or glycerol. Corresponding tandem mass spectrometric analyses detected LamN protein expression only in cells grown on laminarin. Heterologous expression, purification and biochemical characterization identified LamN as a heat stable laminarinase with β-1,3, β-1,4 and β-1,6 glucosidase activity. Collectively, these data identify an enzyme that may allow V. campbellii to exploit some of the most abundant polysaccharides associated with deteriorating phytoplankton blooms and provide support for the potential involvement of V. campbellii in the formation of bioluminescent milky seas.


Frontiers in chemistry | 2014

Development of organophosphate hydrolase activity in a bacterial homolog of human cholinesterase

Patricia M. Legler; Susanne M. Boisvert; Jaimee R. Compton; Charles B. Millard

We applied a combination of rational design and directed evolution (DE) to Bacillus subtilis p-nitrobenzyl esterase (pNBE) with the goal of enhancing organophosphorus acid anhydride hydrolase (OPAAH) activity. DE started with a designed variant, pNBE A107H, carrying a histidine homologous with human butyrylcholinesterase G117H to find complementary mutations that further enhance its OPAAH activity. Five sites were selected (G105, G106, A107, A190, and A400) within a 6.7 Å radius of the nucleophilic serine Oγ. All 95 variants were screened for esterase activity with a set of five substrates: pNP-acetate, pNP-butyrate, acetylthiocholine, butyrylthiocholine, or benzoylthiocholine. A microscale assay for OPAAH activity was developed for screening DE libraries. Reductions in esterase activity were generally concomitant with enhancements in OPAAH activity. One variant, A107K, showed an unexpected 7-fold increase in its kcat/Km for benzoylthiocholine, demonstrating that it is also possible to enhance the cholinesterase activity of pNBE. Moreover, DE resulted in at least three variants with modestly enhanced OPAAH activity compared to wild type pNBE. A107H/A190C showed a 50-fold increase in paraoxonase activity and underwent a slow time- and temperature-dependent change affecting the hydrolysis of OPAA and ester substrates. Structural analysis suggests that pNBE may represent a precursor leading to human cholinesterase and carboxylesterase 1 through extension of two vestigial specificity loops; a preliminary attempt to transfer the Ω-loop of BChE into pNBE is described. Unlike butyrylcholinesterase and pNBE, introducing a G143H mutation (equivalent to G117H) did not confer detectable OP hydrolase activity on human carboxylesterase 1 (hCE1). We discuss the use of pNBE as a surrogate scaffold for the mammalian esterases, and the importance of the oxyanion-hole residues for enhancing the OPAAH activity of selected serine hydrolases.


Biochemistry | 2017

Mutation of Asn-475 in the Venezuelan Equine Encephalitis Virus nsP2 Cysteine Protease leads to a Self-inhibited state

Jaimee R. Compton; Matthew J. Mickey; Xin Hu; Juan J. Marugan; Patricia M. Legler

The alphaviral nsP2 cysteine protease of the Venezuelan equine encephalitis virus (VEEV) is a validated antiviral drug target. Clan CN proteases contain a cysteine protease domain that is intimately packed with an S-adenosyl-l-methionine-dependent RNA methyltransferase (SAM MTase) domain. Within a cleft formed at the interface of these two domains, the peptide substrate is thought to bind. The nucleophilic cysteine can be found within a conserved motif, 475NVCWAK480, which differs from that of papain (22CGSCWAFS29). Mutation of the motif residue, N475, to alanine unexpectedly produced a self-inhibited state in which the N-terminal residues flipped into the substrate-binding cleft. Notably, the N-terminal segment was not hydrolyzed-consistent with a catalytically incompetent state. The N475A mutation resulted in a 70-fold decrease in kcat/Km. A side chain-substrate interaction was predicted by the structure; the S701A mutation led to a 17-fold increase in Km. An Asn at the n-2 position relative to the Cys was also found in the coronaviral papain-like proteases/deubiquitinases (PLpro) of the SARS and MERS viruses, and in several papain-like human ubiquitin specific proteases (USP). The large conformational change in the N475A variant suggests that Asn-475 plays an important role in stabilizing the N-terminal residues and in orienting the carbonyl during nucleophilic attack but does not directly hydrogen bond the oxyanion. The state trapped in crystallo is an unusual result of site-directed mutagenesis but reveals the role of this highly conserved Asn and identifies key substrate-binding contacts that may be exploited by peptide-like inhibitors.

Collaboration


Dive into the Jaimee R. Compton's collaboration.

Top Co-Authors

Avatar

Patricia M. Legler

United States Naval Research Laboratory

View shared research outputs
Top Co-Authors

Avatar

Mark A. Olson

United States Army Medical Research Institute of Infectious Diseases

View shared research outputs
Top Co-Authors

Avatar

Dagmar H. Leary

United States Naval Research Laboratory

View shared research outputs
Top Co-Authors

Avatar

Xin Hu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Charles B. Millard

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Kelly L. Robertson

United States Naval Research Laboratory

View shared research outputs
Top Co-Authors

Avatar

Madhusoodana P. Nambiar

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Richard K. Gordon

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Ajit Jadhav

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Juan J. Marugan

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge